Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Gene ; 724: 144157, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31629820

ABSTRACT

Cellular microRNAs are known to modulate the life-cycle of different viruses. Surprisingly, very little data exists on AAV-induced changes to the cellular microRNAome in general and in hepatic and retinal cells, in particular. We reasoned that inducible microRNA in response to recombinant AAV infection may regulate immediate and long-lived cellular responses necessary for the cell's own survival as well as its ability to control several aspects of viral life-cycle. To study this, we performed a global small RNA sequencing analysis in Adeno-associated virus (AAV) serotypes 2 and 3 infected hepatic and retinal cell models. This screen identified multiple differentially expressed microRNAs, in AAV infected Huh-7 and ARPE-19 cells. Among these, one microRNA (miR-4488) was found to be significantly down regulated (-2.24 fold for AAV2 and -3.32 fold for ARPE-19) in AAV infected cells. An enrichment and pathway analysis of miR-4488 predicted its possible effects on gene targets involved in multiple biological processes including cell-cycle regulation, endoplasmic reticulum stress response and lipid-signalling pathways. Moreover, validation studies in miR-4488 mimic or sponge transfected cells revealed modulation of these target pathways in a cell-specific manner. Further studies demonstrated that overexpression of miR-4488, modestly increased gene expression (126-128%) from AAV2 and AAV3 vectors in Huh-7 cells whereas miR-4488 inhibition in ARPE-19 cells had a similar increase (142-158%) on AAV2 or AAV3 transduction. Our results highlight that recombinant AAV mediated microRNA expression is cell-type and serotype-specific and can target specific host cellular biological pathways.


Subject(s)
Dependovirus/genetics , MicroRNAs/genetics , Parvoviridae Infections/genetics , Retinal Pigment Epithelium/virology , Transduction, Genetic/methods , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Gene Expression Profiling , Humans , Lipid Metabolism/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Parvovirinae/genetics , Reproducibility of Results , Retinal Pigment Epithelium/cytology , Transgenes
3.
Mol Pharm ; 16(11): 4738-4750, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31596095

ABSTRACT

Recombinant adeno-associated virus (AAV)-based gene therapy has been promising, but several host-related transduction or immune challenges remain. For this mode of therapy to be widely applicable, it is crucial to develop high transduction and permeating vectors that infect the target at significantly low doses. Because glycosylation of capsid proteins is known to be rate limiting in the life cycle of many viruses, we reasoned that perturbation of glycosylation sites in AAV2 capsid will enhance gene delivery. In our first set experiments, pharmacological modulation of the glycosylation status in host cells, modestly decreased (1-fold) AAV2 packaging efficacy while it improved their gene expression (∼74%) in vitro. We then generated 24 mutant AAV2 vectors modified to potentially create or disrupt a glycosylation site in its capsid. Three of them demonstrated a 1.3-2.5-fold increase in transgene expression in multiple cell lines (HeLa, Huh7, and ARPE-19). Hepatic gene transfer of these vectors in hemophilia B mice, resulted in a 2-fold increase in human coagulation factor (F)IX levels, while its T/B-cell immunogenic response was unaltered. Subsequently, intravitreal gene transfer of glycosylation site-modified vectors in C57BL6/J mice demonstrated an increase in green fluorescence protein expression (∼2- to 4-fold) and enhanced permeation across retina. Subretinal administration of these modified vectors containing RPE65 gene further rescued the photoreceptor response in a murine model of Leber congenital amarousis. Our studies highlight the translational potential of glycosylation site-modified AAV2 vectors for hepatic and ocular gene therapy applications.


Subject(s)
Capsid Proteins/genetics , Capsid/metabolism , Dependovirus/genetics , Hemophilia A/genetics , Retinal Degeneration/genetics , Animals , Cell Line , Cell Line, Tumor , Disease Models, Animal , Gene Expression/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , HeLa Cells , Hemophilia A/metabolism , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Retina/metabolism , Retinal Degeneration/metabolism , Transduction, Genetic/methods , Transgenes/genetics
4.
Hum Gene Ther ; 30(12): 1461-1476, 2019 12.
Article in English | MEDLINE | ID: mdl-31642343

ABSTRACT

Synthetic engineering of viral vectors such as adeno-associated virus (AAV) is crucial to overcome host transduction barriers observed during clinical gene therapy. We reasoned that exploring the role of cellular ubiquitin-like modifiers (UBLs) such as Neddylation or SUMOylation during AAV transduction could be beneficial. Using a combination of in silico biochemical and molecular engineering strategies, we have studied the impact of these UBLs during AAV2 infection and further developed Neddylation or SUMOylation site-modified AAV vectors and validated them in multiple disease models in vitro and in vivo. Hepatic gene transfer of two novel vectors developed, K105Q (SUMOylation-site mutant) and K665Q (Neddylation-site mutant), demonstrated a significantly improved human coagulation factor (F) IX expression (up to two-fold) in a murine model of hemophilia B. Furthermore, subretinal gene transfer of AAV2-K105Q vector expressing RPE65 gene demonstrated visual correction in a murine model of a retinal degenerative disease (rd12 mice). These vectors did not have any adverse immunogenic events in vivo. Taken together, we demonstrate that gene delivery vectors specifically engineered at UBLs can improve the therapeutic outcome during AAV-mediated ocular or hepatic gene therapy.


Subject(s)
Genetic Therapy , Hemophilia B/therapy , Leber Congenital Amaurosis/therapy , Sumoylation/genetics , Animals , Blood Coagulation Factors/genetics , Dependovirus/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Hemophilia B/genetics , Humans , Leber Congenital Amaurosis/genetics , Mice , Retinal Degeneration/genetics , Retinal Degeneration/therapy
5.
FEBS J ; 286(24): 4964-4981, 2019 12.
Article in English | MEDLINE | ID: mdl-31330090

ABSTRACT

Post-translational modifications in viral capsids are known to fine-tune and regulate several aspects of the infective life cycle of several viruses in the host. Recombinant viruses that are generated in a specific producer cell line are likely to inherit unique post-translational modifications during intra-cellular maturation of its capsid proteins. Data on such post-translational modifications in the capsid of recombinant adeno-associated virus serotypes (AAV1-rh10) is limited. We have employed liquid chromatography and mass spectrometry analysis to characterize post-translational modifications in AAV1-rh10 capsid protein. Our analysis revealed a total of 52 post-translational modifications in AAV2-AAVrh10 capsids, including ubiquitination (17%), glycosylation (36%), phosphorylation (21%), SUMOylation (13%) and acetylation (11%). While AAV1 had no detectable post-translational modification, at least four AAV serotypes had >7 post-translational modifications in their capsid protein. About 82% of these post-translational modifications are novel. A limited validation of AAV2 capsids by MALDI-TOF and western blot analysis demonstrated minimal glycosylation and ubiquitination of AAV2 capsids. To further validate this, we disrupted a glycosylation site identified in AAV2 capsid (AAV2-N253Q), which severely compromised its packaging efficiency (~ 100-fold vs. AAV2 wild-type vectors). In order to confirm other post-translational modifications detected such as SUMOylation, mutagenesis of a SUMOylation site(K258Q) in AAV2 was performed. This mutant vector demonstrated reduced levels of SUMO-1/2/3 proteins and negligible transduction, 2 weeks after ocular gene transfer. Our study underscores the heterogeneity of post-translational modifications in AAV vectors. The data presented here, should facilitate further studies to understand the biological relevance of post-translational modifications in AAV life cycle and the development of novel bioengineered AAV vectors for gene therapy applications. ENZYMES: Trypsin, EC 3.4.21.4.


Subject(s)
Capsid Proteins/metabolism , Parvovirinae/genetics , Acetylation , Capsid Proteins/genetics , Dependovirus , Genetic Vectors/genetics , Glycosylation , HeLa Cells , Humans , Mutation/genetics , Phosphorylation , Protein Processing, Post-Translational , Serogroup , Transduction, Genetic , Ubiquitination
6.
Hum Gene Ther ; 30(8): 921-922, 2019 08.
Article in English | MEDLINE | ID: mdl-31237443

ABSTRACT

We have evaluated the retinal morphological changes in normal C57BL6/J mice and a retinal degenerative disease (rd12 mice) model using scanning electron microscopy. A method of sample preparation for electron microscopy was developed and the cryosectioned retina was used to study the retinal thickness. Our data demonstrate variation in the neural retina texture between rd12 mice and C57BL6/J mice.


Subject(s)
Microscopy, Electron, Scanning , Retina/ultrastructure , Animals , Disease Models, Animal , Electroretinography , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning/methods , Retina/metabolism , Retina/physiopathology , Retinal Degeneration/diagnosis , Retinal Degeneration/pathology
7.
Front Pharmacol ; 8: 441, 2017.
Article in English | MEDLINE | ID: mdl-28769791

ABSTRACT

Of the 12 common serotypes used for gene delivery applications, Adeno-associated virus (AAV)rh.10 serotype has shown sustained hepatic transduction and has the lowest seropositivity in humans. We have evaluated if further modifications to AAVrh.10 at its phosphodegron like regions or predicted immunogenic epitopes could improve its hepatic gene transfer and immune evasion potential. Mutant AAVrh.10 vectors were generated by site directed mutagenesis of the predicted targets. These mutant vectors were first tested for their transduction efficiency in HeLa and HEK293T cells. The optimal vector was further evaluated for their cellular uptake, entry, and intracellular trafficking by quantitative PCR and time-lapse confocal microscopy. To evaluate their potential during hepatic gene therapy, C57BL/6 mice were administered with wild-type or optimal mutant AAVrh.10 and the luciferase transgene expression was documented by serial bioluminescence imaging at 14, 30, 45, and 72 days post-gene transfer. Their hepatic transduction was further verified by a quantitative PCR analysis of AAV copy number in the liver tissue. The optimal AAVrh.10 vector was further evaluated for their immune escape potential, in animals pre-immunized with human intravenous immunoglobulin. Our results demonstrate that a modified AAVrh.10 S671A vector had enhanced cellular entry (3.6 fold), migrate rapidly to the perinuclear region (1 vs. >2 h for wild type vectors) in vitro, which further translates to modest increase in hepatic gene transfer efficiency in vivo. More importantly, the mutant AAVrh.10 vector was able to partially evade neutralizing antibodies (~27-64 fold) in pre-immunized animals. The development of an AAV vector system that can escape the circulating neutralizing antibodies in the host will substantially widen the scope of gene therapy applications in humans.

SELECTION OF CITATIONS
SEARCH DETAIL
...